synaptic vesicle protein 2A

突触囊泡蛋白 2A
  • 文章类型: Journal Article
    目的:突触小泡蛋白2A(SV2A)是药物抵抗癫痫(PRE)的独特治疗靶点。作为癫痫诱发的神经元程序性死亡,pre中很少报道parthanatos。凋亡诱导因子(AIF),与parthanatos有牵连,与SV2A具有共同的细胞保护功能。我们旨在调查parthanatos是否参与PRE并通过AIF通过SV2A缓解。
    方法:采用氯化锂-毛果芸香碱腹腔注射建立癫痫大鼠模型,用苯妥英钠和苯巴比妥钠选择PRE和药敏大鼠。SV2A的表达通过慢病毒递送到海马中进行操作。视频监控用于评估癫痫行为学。在成功的SV2A感染后,采用生化测试来测试海马组织。使用分子动力学计算来模拟SV2A和AIF之间的相互作用。
    结果:Parthanatos核心指数,PARP1,PAR,核AIF和MIF,γ-H2AX,PRE中TUNEL染色均增加。SV2A与AIF结合形成稳定的复合物,成功抑制AIF和MIF核易位和parthanatos,从而减轻PRE的自发性复发性癫痫发作。此外,Parthanatos在SV2A减少后恶化。
    结论:SV2A通过与PRE中的AIF结合来抑制parthanatos,从而保护海马神经元并减轻癫痫发作。
    OBJECTIVE: Synaptic vesicle protein 2A (SV2A) is a unique therapeutic target for pharmacoresistant epilepsy (PRE). As seizure-induced neuronal programmed death, parthanatos was rarely reported in PRE. Apoptosis-inducing factor (AIF), which has been implicated in parthanatos, shares a common cytoprotective function with SV2A. We aimed to investigate whether parthanatos participates in PRE and is mitigated by SV2A via AIF.
    METHODS: An intraperitoneal injection of lithium chloride-pilocarpine was used to establish an epileptic rat model, and phenytoin and phenobarbital sodium were utilized to select PRE and pharmacosensitive rats. The expression of SV2A was manipulated via lentivirus delivery into the hippocampus. Video surveillance was used to assess epileptic ethology. Biochemical tests were employed to test hippocampal tissues following a successful SV2A infection. Molecular dynamic calculations were used to simulate the interaction between SV2A and AIF.
    RESULTS: Parthanatos core index, PARP1, PAR, nuclear AIF and MIF, γ-H2AX, and TUNEL staining were all increased in PRE. SV2A is bound to AIF to form a stable complex, successfully inhibiting AIF and MIF nuclear translocation and parthanatos and consequently mitigating spontaneous recurrent seizures in PRE. Moreover, parthanatos deteriorated after the SV2A reduction.
    CONCLUSIONS: SV2A protected hippocampal neurons and mitigated epileptic seizures by inhibiting parthanatos via binding to AIF in PRE.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    癫痫是一种常见的神经系统疾病,主要用抗癫痫药物(ASM)治疗。虽然诊所里有几十个ASM,大约30%的癫痫患者有药物难治性癫痫发作;大多数传统ASM的其他限制包括耐受性差和药物-药物相互作用.因此,左乙拉西坦(LEV)是一线ASM,耐受性良好,有很好的疗效,几乎没有药物-药物相互作用。尽管人们普遍认为LEV通过独特的治疗靶标突触小泡蛋白(SV)2A起作用,其作用的分子基础仍然未知。即便如此,基于LEV结构的下一代抗癫痫SV2A配体已取得临床成功.本文重点介绍了LEV及其类似物的研发(R&D)过程,brivaracetam和padsevonil,为新型ASM的研发提供思路和经验。
    Epilepsy is a common neurological disorder that is primarily treated with antiseizure medications (ASMs). Although dozens of ASMs are available in the clinic, approximately 30% of epileptic patients have medically refractory seizures; other limitations in most traditional ASMs include poor tolerability and drug-drug interactions. Therefore, there is an urgent need to develop alternative ASMs. Levetiracetam (LEV) is a first-line ASM that is well tolerated, has promising efficacy, and has little drug-drug interaction. Although it is widely accepted that LEV acts through a unique therapeutic target synaptic vesicle protein (SV) 2A, the molecular basis of its action remains unknown. Even so, the next-generation SV2A ligands against epilepsy based on the structure of LEV have achieved clinical success. This review highlights the research and development (R&D) process of LEV and its analogs, brivaracetam and padsevonil, to provide ideas and experience for the R&D of novel ASMs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    本研究探讨了突触小泡蛋白2A(SV2A)在人诱导多能干细胞源性神经干细胞(NSCs)调控中的作用。SV2A在NSC中高度表达。SV2A敲低促进细胞凋亡,这与转录组分析确定的参与p53信号传导的基因上调有关。用小分子p53抑制剂吡虫啉-α治疗逆转了由SV2A缺失诱导的NSC凋亡的促进。这些结果表明SV2A在通过p53信号通路调节NSC存活中起重要作用。
    This study investigated the role of synaptic vesicle protein 2A (SV2A) in the regulation of human induced pluripotent stem cell-derived neural stem cells (NSCs). SV2A was highly expressed in NSCs. SV2A knockdown promotes apoptosis, which was associated with an upregulation of genes involved in p53 signaling as determined by transcriptome analysis. Treatment with the small molecule p53 inhibitor pifithrin-α reversed the promotion of NSC apoptosis induced by loss of SV2A. These results demonstrate that SV2A plays an important role in regulating NSC survival via the p53 signaling pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    探讨布立西坦和左乙拉西坦抗癫痫发作的病理生理机制和不良行为/精神作用。在本研究中,我们使用超高效液相色谱法测定了布立西坦和左乙拉西坦对人工高频振荡(HFO)爆发引起的星形胶质细胞L-谷氨酸释放的影响。此外,采用毛细管免疫印迹法测定了布立西坦和左乙拉西坦对原代培养的大鼠星形胶质细胞质膜中连接蛋白43(Cx43)和突触小泡蛋白2A(SV2A)表达的影响。急性人工快速波纹HFO(500Hz)爆发刺激通过含Cx43的半通道使用依赖性地增加了L-谷氨酸的释放,而不影响质膜中Cx43或SV2A的表达,而急性生理波纹HFO(200Hz)刺激不影响星形胶质细胞L-谷氨酸的释放或Cx43或SV2A的表达。相反,亚慢性波纹HFO和急性病理性快速波纹HFO(500Hz)刺激通过含Cx43的半通道和质膜中Cx43的表达依赖性地增加了L-谷氨酸的释放。亚慢性快速波纹HFO诱发刺激在质膜中产生SV2A的异位表达,但亚慢性波纹HFO刺激未产生异位SV2A。布立西坦和左乙拉西坦的亚慢性给药浓度依赖性地抑制了快速纹波HFO诱导的星形胶质细胞L-谷氨酸的释放以及质膜中Cx43和SV2A的表达。相比之下,亚慢性波纹HFO诱发刺激诱导星形胶质细胞L-谷氨酸释放,和Cx43在质膜中的表达被亚慢性左乙拉西坦给药抑制,但不受brivaracetam的影响。这些结果表明,布立拉西坦和左乙拉西坦抑制了与半通道相关的致癫痫性快速波纹HFO诱导的活化星形胶质细胞传播。相比之下,治疗相关浓度的左乙拉西坦对生理性波纹HFO诱导的星形胶质细胞反应的抑制作用可能与左乙拉西坦的不良行为/精神影响有关.
    To explore the pathophysiological mechanisms of antiseizure and adverse behavioural/psychiatric effects of brivaracetam and levetiracetam, in the present study, we determined the effects of brivaracetam and levetiracetam on astroglial L-glutamate release induced by artificial high-frequency oscillation (HFO) bursts using ultra-high-performance liquid chromatography. Additionally, the effects of brivaracetam and levetiracetam on protein expressions of connexin43 (Cx43) and synaptic vesicle protein 2A (SV2A) in the plasma membrane of primary cultured rat astrocytes were determined using a capillary immunoblotting system. Acutely artificial fast-ripple HFO (500 Hz) burst stimulation use-dependently increased L-glutamate release through Cx43-containing hemichannels without affecting the expression of Cx43 or SV2A in the plasma membrane, whereas acute physiological ripple HFO (200 Hz) stimulation did not affect astroglial L-glutamate release or expression of Cx43 or SV2A. Contrarily, subchronic ripple HFO and acute pathological fast-ripple HFO (500 Hz) stimulations use-dependently increased L-glutamate release through Cx43-containing hemichannels and Cx43 expression in the plasma membrane. Subchronic fast-ripple HFO-evoked stimulation produced ectopic expression of SV2A in the plasma membrane, but subchronic ripple HFO stimulation did not generate ectopic SV2A. Subchronic administration of brivaracetam and levetiracetam concentration-dependently suppressed fast-ripple HFO-induced astroglial L-glutamate release and expression of Cx43 and SV2A in the plasma membrane. In contrast, subchronic ripple HFO-evoked stimulation induced astroglial L-glutamate release, and Cx43 expression in the plasma membrane was inhibited by subchronic levetiracetam administration, but was not affected by brivaracetam. These results suggest that brivaracetam and levetiracetam inhibit epileptogenic fast-ripple HFO-induced activated astroglial transmission associated with hemichannels. In contrast, the inhibitory effect of therapeutic-relevant concentrations of levetiracetam on physiological ripple HFO-induced astroglial responses probably contributes to the adverse behavioural/psychiatric effects of levetiracetam.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    阿尔茨海默病(AD),严重的神经退行性疾病,病理特征为突触丢失和功能障碍。突触囊泡蛋白2A(SV2A)是突触中不可缺少的特异性表达的囊泡蛋白,可作为突触密度的生物标志物。我们发现SV2A在AD患者海马中表达下调,然而,SV2A与AD病理的其他标志如淀粉样前体蛋白(APP)的关系,β-淀粉样蛋白(Aβ),和Tau蛋白不完全清楚。此外,SV2A与APP共定位,在Aβ沉积时下调。此外,我们发现SV2A缺乏导致Aβ和Tau过度磷酸化同时增加,而SV2A过表达与β位点APP裂解酶1和载脂蛋白E基因的下调有关。此外,研究中获得的证据表明,磷脂酰肌醇3-激酶信号通路可能是SV2A调节中影响AD发生和发展的介质.由于AD的有效诊断方法有限,我们的研究表明,SV2A和AD相关蛋白之间的密切相互作用可能提供新颖和创新的诊断和治疗机会.
    Alzheimer\'s disease (AD), a serious neurodegenerative disease, is pathologically characterized by synaptic loss and dysfunction. Synaptic vesicle protein 2A (SV2A) is an indispensable vesicular protein specifically expressed in synapses and can be used as a biomarker for synaptic density. We found that the expression of SV2A was down-regulated in the hippocampus of AD patients, yet the relation of SV2A to other hallmarks of AD pathology such as amyloid precursor protein (APP), β-amyloid (Aβ), and Tau protein is not thoroughly clear. In addition, SV2A colocalized with APP and was down-regulated at Aβ deposition. Moreover, we found that SV2A deficiency leads to a simultaneous increase in Aβ and Tau hyperphosphorylation, while SV2A overexpression was associated with downregulation of β-site APP cleaving enzyme 1 and apolipoprotein E genes. In addition, evidence gained in the study points to the phosphatidylinositol 3-kinase signaling pathway as a possible mediator in SV2A regulation influencing the incidence and development of AD. With limited effective diagnostic methods for AD, a close interplay between SV2A and AD-related proteins demonstrated in our study may provide novel and innovative diagnostic and therapeutic opportunities.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    The antiepileptic/anticonvulsive action of brivaracetam is considered to occur via modulation of synaptic vesicle protein 2A (SV2A); however, the pharmacological mechanisms of action have not been fully characterised. To explore the antiepileptic/anticonvulsive mechanism of brivaracetam associated with SV2A modulation, this study determined concentration-dependent effects of brivaracetam on astroglial L-glutamate release associated with connexin43 (Cx43), tumour-necrosis factor-α (TNFα) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/glutamate receptor of rat primary cultured astrocytes using ultra-high-performance liquid chromatography. Furthermore, interaction among TNFα, elevated extracellular K+ and brivaracetam on expression of SV2A and Cx43 was determined using capillary immunoblotting. TNFα and elevated extracellular K+ predominantly enhanced astroglial L-glutamate release associated with respective AMPA/glutamate receptor and hemichannel. These effects were enhanced by a synergistic effect of TNFα and elevated extracellular K+ in combination. The activation of astroglial L-glutamate release, and expression of SV2A and Cx43 in the plasma membrane was suppressed by subchronic brivaracetam administration but were unaffected by acute administration. These results suggest that migration of SV2A to the astroglial plasma membrane by hyperexcitability activates astroglial glutamatergic transmission, perhaps via hemichannel activation. Subchronic brivaracetam administration suppressed TNFα-induced activation of AMPA/glutamate receptor and hemichannel via inhibition of ectopic SV2A. These findings suggest that combined inhibition of vesicular and ectopic SV2A functions contribute to the antiepileptic/anticonvulsive mechanism of brivaracetam action.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    突触丢失是许多神经退行性疾病的突出和早期特征。
    我们测试了以下假设:进行性核上性麻痹(PSP)(Richardson综合征)和淀粉样蛋白阴性皮质基底综合征(CBS)的原发性tau蛋白病变中突触密度降低。
    44名参与者(15名CBS,14PSP,和15个年龄/性别/教育匹配的对照)用放射性配体[11C]UCB-J进行PET,与突触囊泡糖蛋白2A结合,突触密度的标志物;参与者还进行了3TeslaMRI以及临床和神经心理学评估。
    九名CBS患者具有通过[11C]PiBPET测定的阴性淀粉样蛋白生物标志物,因此被认为可能具有皮质基底变性(CBD)。PSP-Richardson综合征和淀粉样蛋白阴性CBS的患者在执行方面受损,记忆,和视觉空间任务。[11C]UCB-J结合在额叶上减少,temporal,顶叶,和枕叶,扣带回,海马体,脑岛,杏仁核,与对照组相比,PSP和CBD患者的皮质下结构(P<0.01),中位数下降高达50%,与验尸数据一致。即使在萎缩程度最小的大脑区域,也普遍减少20%至30%。总体[11C]UCB-J结合与PSP和CBD评分量表之间呈负相关(R=-0.61,P<0.002;R=-0.72,P<0.001),与修订后的Addenbrooke认知检查呈正相关(R=0.52;P=0.01)。
    我们确认PSP和CBD的严重突触丢失与疾病严重程度成正比,提供对原发性退行性tau蛋白病的病理生理学的关键见解。[11C]UCB-J可以促进疾病修饰的治疗策略,突触维持,或恢复。©2020作者由WileyPeriodicalsLLC代表国际帕金森症和运动障碍协会出版的运动障碍。
    Synaptic loss is a prominent and early feature of many neurodegenerative diseases.
    We tested the hypothesis that synaptic density is reduced in the primary tauopathies of progressive supranuclear palsy (PSP) (Richardson\'s syndrome) and amyloid-negative corticobasal syndrome (CBS).
    Forty-four participants (15 CBS, 14 PSP, and 15 age-/sex-/education-matched controls) underwent PET with the radioligand [11 C]UCB-J, which binds to synaptic vesicle glycoprotein 2A, a marker of synaptic density; participants also had 3 Tesla MRI and clinical and neuropsychological assessment.
    Nine CBS patients had negative amyloid biomarkers determined by [11 C]PiB PET and hence were deemed likely to have corticobasal degeneration (CBD). Patients with PSP-Richardson\'s syndrome and amyloid-negative CBS were impaired in executive, memory, and visuospatial tasks. [11 C]UCB-J binding was reduced across frontal, temporal, parietal, and occipital lobes, cingulate, hippocampus, insula, amygdala, and subcortical structures in both PSP and CBD patients compared to controls (P < 0.01), with median reductions up to 50%, consistent with postmortem data. Reductions of 20% to 30% were widespread even in areas of the brain with minimal atrophy. There was a negative correlation between global [11 C]UCB-J binding and the PSP and CBD rating scales (R = -0.61, P < 0.002; R = -0.72, P < 0.001, respectively) and a positive correlation with the revised Addenbrooke\'s Cognitive Examination (R = 0.52; P = 0.01).
    We confirm severe synaptic loss in PSP and CBD in proportion to disease severity, providing critical insight into the pathophysiology of primary degenerative tauopathies. [11 C]UCB-J may facilitate treatment strategies for disease-modification, synaptic maintenance, or restoration. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    11C-UCB-J((R)-1-((3-(11C-甲基-11C)吡啶-4-基)甲基)-4-(3,4,5-三氟苯基)吡咯烷-2-酮)是突触小泡糖蛋白2A的PET示踪剂,这可能是突触密度的标志。为了简化扫描协议,将SUV比率(SUVR)与基于模型的非位移结合电位(BPND)进行比较,以选择健康和神经精神受试者的最佳时间窗口。方法:总计,在90分钟内获得141次扫描。进行了动脉血采样和代谢物分析。在六个30分钟的窗口中计算SUVR-1(半卵中心参考区域),并与1-组织室模型BPND进行比较。进行模拟以评估SUVR-1的时间依赖性。结果:对于所有受试者,在较晚的时间窗口观察到SUVR-1更大的相关性和更少的偏倚。仿真结果表明,SUVR-1和BPND之间的协议是时间依赖性的。结论:60至90分钟的时间段提供了SUVR-1和BPND之间的最佳匹配(-1%±7%);因此,短扫描足以准确定量11C-UCB-J特异性结合。
    11C-UCB-J ((R)-1-((3-(11C-methyl-11C)pyridin-4-yl)methyl)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one) is a PET tracer for synaptic vesicle glycoprotein 2A, which may be a marker of synaptic density. To simplify the scan protocol, SUV ratios (SUVRs) were compared with model-based nondisplaceable binding potential (BPND) to select the optimal time window in healthy and neuropsychiatric subjects. Methods: In total, 141 scans were acquired for 90 min. Arterial blood sampling and metabolite analysis were conducted. SUVR-1 (centrum semiovale reference region) was computed for six 30-min windows and compared with 1-tissue-compartment model BPND Simulations were performed to assess the time dependency of SUVR-1. Results: Greater correlation and less bias were observed for SUVR-1 at later time windows for all subjects. Simulations showed that the agreement between SUVR-1 and BPND is time-dependent. Conclusion: The 60- to 90-min period provided the best match between SUVR-1 and BPND (-1% ± 7%); thus, a short scan is sufficient for accurate quantification of 11C-UCB-J-specific binding.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    颞叶癫痫(TLE)患者通常患有认知障碍和反复发作。布立西坦(BRV)是一种新型的抗癫痫药物(AEDs),最近用于治疗有或没有继发性推广的部分性癫痫发作。与其他AED不同,BRV在突触可塑性方面具有一些有利的性质。然而,潜在的机制仍然难以捉摸。
    本研究旨在探讨BRV对实验性TLE大鼠突触可塑性的神经保护机制。
    通过体内场兴奋性突触后电位(fEPSP)的测量,在毛果芸香碱诱导的TLE模型上评估了BRV(10mg/kg)慢性治疗的效果。用免疫印迹法鉴定了差异表达的突触囊泡蛋白2A(SV2A)。然后,在TLE模型中长时程增强(LTP)诱导过程中,突触体相关蛋白25(SNAP-25)的快速磷酸化研究了BRV对突触可塑性的潜在作用.
    在癫痫大鼠中显示出海马中SV2A水平的升高伴随着LTP的降低。此外,BRV治疗持续30天以上,改善了癫痫大鼠SV2A的过表达,逆转了突触功能障碍。此外,BRV治疗减轻了癫痫患者LTP诱导过程中Ser187上SNAP-25的异常磷酸化,这与突触小泡胞吐和电压门控钙通道的调节有关。
    BRV治疗改善了癫痫大鼠海马中SV2A的过度表达并挽救了突触功能障碍。这些结果确定了BRV对TLE模型的神经保护作用。
    Patients with temporal lobe epilepsy (TLE) usually suffer from cognitive deficits and recurrent seizures. Brivaracetam (BRV) is a novel anti-epileptic drug (AEDs) recently used for the treatment of partial seizures with or without secondary generalization. Different from other AEDs, BRV has some favorable properties on synaptic plasticity. However, the underlying mechanisms remain elusive.
    The aim of this study was to explore the neuroprotective mechanism of BRV on synaptic plasticity in experimental TLE rats.
    The effect of chronic treatment with BRV (10 mg/kg) was assessed on Pilocarpine induced TLE model through measurement of the field excitatory postsynaptic potentials (fEPSPs) in vivo. Differentially expressed synaptic vesicle protein 2A (SV2A) were identified with immunoblot. Then, fast phosphorylation of synaptosomal-associated protein 25 (SNAP-25) during long-term potentiation (LTP) induction was performed to investigate the potential roles of BRV on synaptic plasticity in the TLE model.
    An increased level of SV2A accompanied by a depressed LTP in the hippocampus was shown in epileptic rats. Furthermore, BRV treatment continued for more than 30 days improved the over-expression of SV2A and reversed the synaptic dysfunction in epileptic rats. Additionally, BRV treatment alleviates the abnormal SNAP-25 phosphorylation at Ser187 during LTP induction in epileptic ones, which is relevant to the modulation of synaptic vesicles exocytosis and voltagegated calcium channels.
    BRV treatment ameliorated the over-expression of SV2A in the hippocampus and rescued the synaptic dysfunction in epileptic rats. These results identify the neuroprotective effect of BRV on TLE model.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    使用老年抗癫痫药物(AEDs)的长期治疗,但不是左乙拉西坦(LEV),可能会增加动脉粥样硬化(AS)的风险,提示LEV可能具有潜在的抗AS作用。已知突触小泡2A(SV2A)是LEV的特异性结合位点。大量研究表明,SV2A是一种在神经系统中特异性表达的膜蛋白。有趣的是,我们之前的研究表明SV2A也存在于人类CD8+T淋巴细胞中。因此,我们假设LEV通过调节单核细胞趋化和粘附而降低AS风险.我们显示在THP-1人单核细胞白血病细胞中检测到SV2A蛋白。LEV(300μM)抑制表达SV2AWT的质粒转染后THP-1细胞的趋化和粘附,但不是SV2AR383Q,这是人类SV2A的已知功能突变位点。此外,RT-PCR和westernblot分析表明,LEV(300μM)降低趋化因子相关受体(CX3CL1,CCR1,CCR2和CCR5)的表达水平,用SV2AWT表达质粒降低THP-1细胞中磷酸化AKT(p-AKT)的水平。一起来看,这些发现表明LEV对THP-1单核细胞粘附和趋化具有抑制作用,提示SV2A可能成为预防AS的新治疗靶点。
    Long-term therapy with older antiepileptic drugs (AEDs), but not levetiracetam (LEV), may increase the risk of atherosclerosis (AS), suggesting that LEV may have a potential anti-AS effect. The synaptic vesicle 2A (SV2A) is known to the specific binding site of LEV. Numerous studies have documented that SV2A is a membrane protein specifically expressed in nervous system. Interestingly, our previous research showed that SV2A also existed in human CD8+ T lymphocytes. Therefore, we hypothesized that LEV was associated with decreased risk of AS by regulating monocytes chemotaxis and adhesion. We showed that SV2A protein were detected in THP-1 human monocytic leukemia cells. LEV (300 μM) inhibited the chemotaxis and adhesion of THP-1 cells after transfection with plasmids expressing SV2AWT, but not SV2AR383Q which was a known functional mutation site of human SV2A. Furthermore, RT-PCR and western blot analysis demonstrated that LEV (300 μM) decreased the expression level of chemokine-related receptors (CX3CL1, CCR1, CCR2, and CCR5),and reduced levels of phosphorylated AKT (p-AKT) in THP-1 cells with SV2AWT expressing plasmids. Taken together, these findings indicated that LEV has an inhibitory effect on THP-1 monocyte adhesion and chemotaxis, suggesting that SV2A may serve as a novel therapeutic target to prevent AS.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号